Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Proc Natl Acad Sci U S A ; 120(37): e2305494120, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37669364

ABSTRACT

Cryoelectron microscopy (Cryo-EM) has enabled structural determination of proteins larger than about 50 kDa, including many intractable by any other method, but it has largely failed for smaller proteins. Here, we obtain structures of small proteins by binding them to a rigid molecular scaffold based on a designed protein cage, revealing atomic details at resolutions reaching 2.9 Å. We apply this system to the key cancer signaling protein KRAS (19 kDa in size), obtaining four structures of oncogenic mutational variants by cryo-EM. Importantly, a structure for the key G12C mutant bound to an inhibitor drug (AMG510) reveals significant conformational differences compared to prior data in the crystalline state. The findings highlight the promise of cryo-EM scaffolds for advancing the design of drug molecules against small therapeutic protein targets in cancer and other human diseases.


Subject(s)
Diagnostic Imaging , Humans , Cryoelectron Microscopy
2.
J Med Chem ; 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080842

ABSTRACT

PRMT5, a type 2 arginine methyltransferase, has a critical role in regulating cell growth and survival in cancer. With the aim of developing MTA-cooperative PRMT5 inhibitors suitable for MTAP-deficient cancers, herein we report our efforts to develop novel "MTA-cooperative" compounds identified through a high-throughput biochemical screening approach. Optimization of hits was achieved through structure-based design with a focus on improvement of oral drug-like properties. Bioisosteric replacement of the original thiazole guanidine headgroup, spirocyclization of the isoindolinone amide scaffold to both configurationally and conformationally lock the bioactive form, and fine-tuning of the potency, MTA cooperativity, and DMPK properties through specific substitutions of the azaindole headgroup were conducted. We have identified an orally available in vivo lead compound, 28 ("AZ-PRMT5i-1"), which shows sub-10 nM PRMT5 cell potency, >50-fold MTA cooperativity, suitable DMPK properties for oral dosing, and significant PRMT5-driven in vivo efficacy in several MTAP-deficient preclinical cancer models.

3.
Acta Crystallogr D Struct Biol ; 79(Pt 6): 449-461, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37259835

ABSTRACT

The Collaborative Computational Project No. 4 (CCP4) is a UK-led international collective with a mission to develop, test, distribute and promote software for macromolecular crystallography. The CCP4 suite is a multiplatform collection of programs brought together by familiar execution routines, a set of common libraries and graphical interfaces. The CCP4 suite has experienced several considerable changes since its last reference article, involving new infrastructure, original programs and graphical interfaces. This article, which is intended as a general literature citation for the use of the CCP4 software suite in structure determination, will guide the reader through such transformations, offering a general overview of the new features and outlining future developments. As such, it aims to highlight the individual programs that comprise the suite and to provide the latest references to them for perusal by crystallographers around the world.


Subject(s)
Proteins , Software , Proteins/chemistry , Crystallography, X-Ray , Macromolecular Substances
4.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 4): 425-30, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22505262

ABSTRACT

Coot is a molecular-graphics application primarily aimed to assist in model building and validation of biological macromolecules. Recently, tools have been added to work with small molecules. The newly incorporated tools for the manipulation and validation of ligands include interaction with PRODRG, subgraph isomorphism-based tools, representation of ligand chemistry, ligand fitting and analysis, and are described here.


Subject(s)
Crystallography, X-Ray/methods , Ligands , Models, Molecular , Software
5.
EMBO J ; 27(13): 1907-18, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18566585

ABSTRACT

The positive transcription elongation factor b (P-TEFb) (CDK9/cyclin T (CycT)) promotes mRNA transcriptional elongation through phosphorylation of elongation repressors and RNA polymerase II. To understand the regulation of a transcriptional CDK by its cognate cyclin, we have determined the structures of the CDK9/CycT1 and free cyclin T2. There are distinct differences between CDK9/CycT1 and the cell cycle CDK CDK2/CycA manifested by a relative rotation of 26 degrees of CycT1 with respect to the CDK, showing for the first time plasticity in CDK cyclin interactions. The CDK9/CycT1 interface is relatively sparse but retains some core CDK-cyclin interactions. The CycT1 C-terminal helix shows flexibility that may be important for the interaction of this region with HIV TAT and HEXIM. Flavopiridol, an anticancer drug in phase II clinical trials, binds to the ATP site of CDK9 inducing unanticipated structural changes that bury the inhibitor. CDK9 activity and recognition of regulatory proteins are governed by autophosphorylation. We show that CDK9/CycT1 autophosphorylates on Thr186 in the activation segment and three C-terminal phosphorylation sites. Autophosphorylation on all sites occurs in cis.


Subject(s)
Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/metabolism , Cyclins/metabolism , Flavonoids/metabolism , Piperidines/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cyclin T , Cyclins/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Sequence Alignment
6.
Structure ; 17(3): 352-62, 2009 Mar 11.
Article in English | MEDLINE | ID: mdl-19278650

ABSTRACT

Splicing requires reversible phosphorylation of serine/arginine-rich (SR) proteins, which direct splice site selection in eukaryotic mRNA. These phosphorylation events are dependent on SR protein (SRPK) and cdc2-like kinase (CLK) families. SRPK1 phosphorylation of splicing factors is restricted by a specific docking interaction whereas CLK activity is less constrained. To understand functional differences between splicing factor targeting kinases, we determined crystal structures of CLK1 and CLK3. Intriguingly, in CLKs the SRPK1 docking site is blocked by insertion of a previously unseen helix alphaH. In addition, substrate docking grooves present in related mitogen activating protein kinases (MAPKs) are inaccessible due to a CLK specific beta7/8-hairpin insert. Thus, the unconstrained substrate interaction together with the determined active-site mediated substrate specificity allows CLKs to complete the functionally important hyperphosphorylation of splicing factors like ASF/SF2. In addition, despite high sequence conservation, we identified inhibitors with surprising isoform specificity for CLK1 over CLK3.


Subject(s)
Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Binding Sites , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Insertional , Nuclear Proteins/chemistry , Phosphorylation , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , RNA Splicing , RNA-Binding Proteins/chemistry , Serine-Arginine Splicing Factors , Substrate Specificity
7.
SLAS Discov ; 26(1): 17-31, 2021 01.
Article in English | MEDLINE | ID: mdl-33016175

ABSTRACT

Single-particle cryogenic electron microscopy (cryo-EM) has been elevated to the mainstream of structural biology propelled by technological advancements in numerous fronts, including imaging analysis and the development of direct electron detectors. The drug discovery field has watched with (initial) skepticism and wonder at the progression of the technique and how it revolutionized the molecular understanding of previously intractable targets. This article critically assesses how cryo-EM has impacted drug discovery in diverse therapeutic areas. Targets that have been brought into the realm of structure-based drug design by cryo-EM and are thus reviewed here include membrane proteins like the GABAA receptor, several TRP channels, and G protein-coupled receptors, and multiprotein complexes like the ribosomes, the proteasome, and eIF2B. We will describe these studies highlighting the achievements, challenges, and caveats.


Subject(s)
Cryoelectron Microscopy/methods , Drug Discovery/methods , Animals , Humans , Structure-Activity Relationship
8.
Structure ; 16(1): 115-24, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18184589

ABSTRACT

The activation segment of protein kinases is structurally highly conserved and central to regulation of kinase activation. Here we report an atypical activation segment architecture in human MPSK1 comprising a beta sheet and a large alpha-helical insertion. Sequence comparisons suggested that similar activation segments exist in all members of the MPSK1 family and in MAST kinases. The consequence of this nonclassical activation segment on substrate recognition was studied using peptide library screens that revealed a preferred substrate sequence of X-X-P/V/I-phi-H/Y-T*-N/G-X-X-X (phi is an aliphatic residue). In addition, we identified the GTPase DRG1 as an MPSK1 interaction partner and specific substrate. The interaction domain in DRG1 was mapped to the N terminus, leading to recruitment and phosphorylation at Thr100 within the GTPase domain. The presented data reveal an atypical kinase structural motif and suggest a role of MPSK1 regulating DRG1, a GTPase involved in regulation of cellular growth.


Subject(s)
Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Conserved Sequence , Enzyme Activation , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Staurosporine/metabolism , Substrate Specificity
9.
Structure ; 15(11): 1493-504, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17997974

ABSTRACT

Tyrosine kinase signaling is tightly controlled by negative feedback inhibitors including suppressors of cytokine signaling (SOCS). SOCS assemble as SH2 domain substrate recognition modules in ElonginB/C-cullin ubiquitin ligases. In accordance, SOCS4 reduces STAT3 signaling from EGFR through increased receptor degradation. Variable C-termini in SOCS4-SOCS7 exclude these family members from a SOCS2-type domain arrangement in which a strictly conserved C terminus determines domain packing. The structure of the SOCS4-ElonginC-ElonginB complex reveals a distinct SOCS structural class. The N-terminal ESS helix functionally replaces the CIS/SOCS1-SOCS3 family C terminus in a distinct SH2-SOCS box interface that facilitates further interdomain packing between the extended N- and C-terminal regions characteristic for this subfamily. Using peptide arrays and calorimetry the STAT3 site in EGFR (pY(1092)) was identified as a high affinity SOCS4 substrate (K(D) = 0.5 microM) revealing a mechanism for EGFR degradation. SOCS4 also bound JAK2 and KIT with low micromolar affinity, whereas SOCS2 was specific for GH-receptor.


Subject(s)
ErbB Receptors/metabolism , Suppressor of Cytokine Signaling Proteins/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Elongin , Humans , Isoleucine/metabolism , Models, Molecular , Molecular Sequence Data , Phosphotyrosine/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Sequence Alignment , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factors/metabolism
10.
Structure ; 15(2): 201-13, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17292838

ABSTRACT

p21-activated kinases have been classified into two groups based on their domain architecture. Group II PAKs (PAK4-6) regulate a wide variety of cellular functions, and PAK deregulation has been linked to tumor development. Structural comparison of five high-resolution structures comprising all active, monophosphorylated group II catalytic domains revealed a surprising degree of domain plasticity, including a number of catalytically productive and nonproductive conformers. Rearrangements of helix alphaC, a key regulatory element of kinase function, resulted in an additional helical turn at the alphaC N terminus and a distortion of its C terminus, a movement hitherto unseen in protein kinases. The observed structural changes led to the formation of interactions between conserved residues that structurally link the glycine-rich loop, alphaC, and the activation segment and firmly anchor alphaC in an active conformation. Inhibitor screening identified six potent PAK inhibitors from which a tri-substituted purine inhibitor was cocrystallized with PAK4 and PAK5.


Subject(s)
Catalytic Domain , Protein Serine-Threonine Kinases/chemistry , Amino Acid Sequence , Animals , Catalytic Domain/drug effects , Catalytic Domain/genetics , Crystallography , Molecular Sequence Data , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Purines/chemistry
11.
MAbs ; 10(1): 104-117, 2018 01.
Article in English | MEDLINE | ID: mdl-28952876

ABSTRACT

C5a is a potent anaphylatoxin that modulates inflammation through the C5aR1 and C5aR2 receptors. The molecular interactions between C5a-C5aR1 receptor are well defined, whereas C5a-C5aR2 receptor interactions are poorly understood. Here, we describe the generation of a human antibody, MEDI7814, that neutralizes C5a and C5adesArg binding to the C5aR1 and C5aR2 receptors, without affecting complement-mediated bacterial cell killing. Unlike other anti-C5a mAbs described, this antibody has been shown to inhibit the effects of C5a by blocking C5a binding to both C5aR1 and C5aR2 receptors. The crystal structure of the antibody in complex with human C5a reveals a discontinuous epitope of 22 amino acids. This is the first time the epitope for an antibody that blocks C5aR1 and C5aR2 receptors has been described, and this work provides a basis for molecular studies aimed at further understanding the C5a-C5aR2 receptor interaction. MEDI7814 has therapeutic potential for the treatment of acute inflammatory conditions in which both C5a receptors may mediate inflammation, such as sepsis or renal ischemia-reperfusion injury.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody Affinity , Complement C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptors, Chemokine/antagonists & inhibitors , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibody Specificity , Binding Sites, Antibody , Complement C5a/chemistry , Complement C5a/immunology , Complement C5a/metabolism , Epitope Mapping/methods , Epitopes , HEK293 Cells , Humans , Protein Binding , Protein Conformation , Protein Engineering , Receptor, Anaphylatoxin C5a/chemistry , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Chemokine/chemistry , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Structure-Activity Relationship
12.
Biochem J ; 395(3): 483-91, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16441242

ABSTRACT

Protein tyrosine phosphatases PTPN5, PTPRR and PTPN7 comprise a family of phosphatases that specifically inactivate MAPKs (mitogen-activated protein kinases). We have determined high-resolution structures of all of the human family members, screened them against a library of 24000 compounds and identified two classes of inhibitors, cyclopenta[c]quinolinecarboxylic acids and 2,5-dimethylpyrrolyl benzoic acids. Comparative structural analysis revealed significant differences within this conserved family that could be explored for the design of selective inhibitors. PTPN5 crystallized, in two distinct crystal forms, with a sulphate ion in close proximity to the active site and the WPD (Trp-Pro-Asp) loop in a unique conformation, not seen in other PTPs, ending in a 3(10)-helix. In the PTPN7 structure, the WPD loop was in the closed conformation and part of the KIM (kinase-interaction motif) was visible, which forms an N-terminal aliphatic helix with the phosphorylation site Thr66 in an accessible position. The WPD loop of PTPRR was open; however, in contrast with the structure of its mouse homologue, PTPSL, a salt bridge between the conserved lysine and aspartate residues, which has been postulated to confer a more rigid loop structure, thereby modulating activity in PTPSL, does not form in PTPRR. One of the identified inhibitor scaffolds, cyclopenta[c]quinoline, was docked successfully into PTPRR, suggesting several possibilities for hit expansion. The determined structures together with the established SAR (structure-activity relationship) propose new avenues for the development of selective inhibitors that may have therapeutic potential for treating neurodegenerative diseases in the case of PTPRR or acute myeloblastic leukaemia targeting PTPN7.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/classification , Ions/chemistry , Ions/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/classification , Protein Tyrosine Phosphatases, Non-Receptor , Receptor-Like Protein Tyrosine Phosphatases, Class 7 , Sequence Alignment , Sequence Homology, Amino Acid , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity
13.
J Med Chem ; 60(8): 3438-3450, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28376306

ABSTRACT

There are a number of small-molecule inhibitors targeting the RAS/RAF/MEK/ERK signaling pathway that have either been approved or are in clinical development for oncology across a range of disease indications. The inhibition of ERK1/2 is of significant current interest, as cell lines with acquired resistance to BRAF and MEK inhibitors have been shown to maintain sensitivity to ERK1/2 inhibition in preclinical models. This article reports on our recent work to identify novel, potent, and selective reversible ERK1/2 inhibitors from a low-molecular-weight, modestly active, and highly promiscuous chemical start point, compound 4. To guide and inform the evolution of this series, inhibitor binding mode information from X-ray crystal structures was critical in the rapid exploration of this template to compound 35, which was active when tested in in vivo antitumor efficacy experiments.


Subject(s)
MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Biological Availability , Cell Line, Tumor , Dogs , Drug Discovery , Humans , Methylation , Protein Kinase Inhibitors/pharmacokinetics
14.
Protein Sci ; 15(6): 1500-5, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16672235

ABSTRACT

The receptor-type protein tyrosine phosphatases (RPTPs) are integral membrane proteins composed of extracellular adhesion molecule-like domains, a single transmembrane domain, and a cytoplasmic domain. The cytoplasmic domain consists of tandem PTP domains, of which the D1 domain is enzymatically active. RPTPkappa is a member of the R2A/IIb subfamily of RPTPs along with RPTPmu, RPTPrho, and RPTPlambda. Here, we have determined the crystal structure of catalytically active, monomeric D1 domain of RPTPkappa at 1.9 A. Structural comparison with other PTP family members indicates an overall classical PTP architecture of twisted mixed beta-sheets flanked by alpha-helices, in which the catalytically important WPD loop is in an unhindered open conformation. Though the residues forming the dimeric interface in the RPTPmu structure are all conserved, they are not involved in the protein-protein interaction in RPTPkappa. The N-terminal beta-strand, formed by betax association with betay, is conserved only in RPTPs but not in cytosolic PTPs, and this feature is conserved in the RPTPkappa structure forming a beta-strand. Analytical ultracentrifugation studies show that the presence of reducing agents and higher ionic strength are necessary to maintain RPTPkappa as a monomer. In this family the crystal structure of catalytically active RPTPmu D1 was solved as a dimer, but the dimerization was proposed to be a consequence of crystallization since the protein was monomeric in solution. In agreement, we show that RPTPkappa is monomeric in solution and crystal structure.


Subject(s)
Protein Tyrosine Phosphatases/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Sequence Homology, Amino Acid , Solutions , Structural Homology, Protein
15.
ACS Med Chem Lett ; 7(5): 514-9, 2016 May 12.
Article in English | MEDLINE | ID: mdl-27190603

ABSTRACT

A novel series of covalent inhibitors of EGFR (epidermal growth factor receptor) kinase was discovered through a combination of subset screening and structure-based design. These compounds preferentially inhibit mutant forms of EGFR (activating mutant and T790M mutant) over wild-type EGFR in cellular assays measuring EGFR autophosphorylation and proliferation, suggesting an improved therapeutic index in non-small cell lung cancer patients would be achievable relative to established EGFR inhibitors. We describe our design approaches, resulting in the identification of the lead compound 5, and our efforts to develop an understanding of the structure-activity relationships within this series. In addition, strategies to overcome challenges around metabolic stability and aqueous solubility are discussed. Despite limitations in its physical properties, 5 is orally bioavailable in mice and demonstrates pronounced antitumor activity in in vivo models of mutant EGFR-driven cancers.

16.
J Med Chem ; 48(24): 7604-14, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16302800

ABSTRACT

The kinase PIM-1 plays a pivotal role in cytokine signaling and is implicated in the development of a number of tumors. The three-dimensional structure of PIM-1 is characterized by an unique hinge region which lacks a second hydrogen bond donor and makes it particularly important to determine how inhibitors bind to this kinase. We determined the structures of PIM-1 in complex with bisindolylmaleimide (BIM-1) and established the structure-activity relationship (SAR) for this inhibitor class. In addition, we screened a kinase targeted library and identified a number of high affinity inhibitors of PIM-1 such as imidazo[1,2-b]pyridazines, pyrazolo[1,5-a]pyrimidines, and members of the flavonoid family. In this paper we present an initial SAR of the identified scaffolds determined on the basis of a thermostability shift assay, calorimetric binding data, and biochemical assays which may find applications for the treatment of PIM-1 dependent cancer types.


Subject(s)
Indoles/chemical synthesis , Maleimides/chemical synthesis , Moloney murine leukemia virus/physiology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/chemistry , Proviruses/physiology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Crystallization , Crystallography, X-Ray , Flavonoids/chemical synthesis , Flavonoids/chemistry , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Indoles/chemistry , Maleimides/chemistry , Models, Molecular , Molecular Structure , Moloney murine leukemia virus/enzymology , Phosphorylation , Protein Binding , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyridazines/chemical synthesis , Pyridazines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
17.
J Med Chem ; 58(1): 278-93, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25255283

ABSTRACT

Two structurally distinct series of novel, MAPK-activated kinase-2 prevention of activation inhibitors have been discovered by high throughput screening. Preliminary structure-activity relationship (SAR) studies revealed substructural features that influence the selective inhibition of the activation by p38α of the downstream kinase MK2 in preference to an alternative substrate, MSK1. Enzyme kinetics, surface plasmon resonance (SPR), 2D protein NMR, and X-ray crystallography were used to determine the binding mode and the molecular mechanism of action. The compounds bind competitively to the ATP binding site of p38α but unexpectedly with higher affinity in the p38α-MK2 complex compared with p38α alone. This observation is hypothesized to be the origin of the substrate selectivity. The two lead series identified are suitable for further investigation for their potential to treat chronic inflammatory diseases with improved tolerability over previously studied p38α inhibitors.


Subject(s)
MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Binding, Competitive , Cells, Cultured , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Discovery , Enzyme Activation/drug effects , Humans , Kinetics , MAP Kinase Kinase 2/metabolism , Magnetic Resonance Spectroscopy , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/chemistry , Mitogen-Activated Protein Kinase 14/metabolism , Models, Chemical , Models, Molecular , Molecular Structure , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Structure-Activity Relationship , Substrate Specificity , Surface Plasmon Resonance
18.
J Med Chem ; 58(11): 4790-801, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25977981

ABSTRACT

The RAS/RAF/MEK/ERK signaling pathway has been targeted with a number of small molecule inhibitors in oncology clinical development across multiple disease indications. Importantly, cell lines with acquired resistance to B-RAF and MEK inhibitors have been shown to maintain sensitivity to ERK1/2 inhibition by small molecule inhibitors. There are a number of selective, noncovalent ERK1/2 inhibitors reported along with the promiscuous hypothemycin (and related analogues) that act via a covalent mechanism of action. This article reports the identification of multiple series of highly selective covalent ERK1/2 inhibitors informed by structure-based drug design (SBDD). As a starting point for these covalent inhibitors, reported ERK1/2 inhibitors and a chemical series identified via high-throughput screening were exploited. These approaches resulted in the identification of selective covalent tool compounds for potential in vitro and in vivo studies to assess the risks and or benefits of targeting this pathway through such a mechanism of action.


Subject(s)
Drug Design , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 3/chemistry , Protein Kinase Inhibitors/pharmacology , Amino Acid Sequence , Cells, Cultured , Crystallography, X-Ray , Humans , Immunoblotting , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
19.
Protein Sci ; 23(5): 627-38, 2014 May.
Article in English | MEDLINE | ID: mdl-24677421

ABSTRACT

The EphB receptors have key roles in cell morphology, adhesion, migration and invasion, and their aberrant action has been linked with the development and progression of many different tumor types. Their conflicting expression patterns in cancer tissues, combined with their high sequence and structural identity, present interesting challenges to those seeking to develop selective therapeutic molecules targeting this large receptor family. Here, we present the first structure of the EphB1 tyrosine kinase domain determined by X-ray crystallography to 2.5Å. Our comparative crystalisation analysis of the human EphB family kinases has also yielded new crystal forms of the human EphB2 and EphB4 catalytic domains. Unable to crystallize the wild-type EphB3 kinase domain, we used rational engineering (based on our new structures of EphB1, EphB2, and EphB4) to identify a single point mutation which facilitated its crystallization and structure determination to 2.2 Å. This mutation also improved the soluble recombinant yield of this kinase within Escherichia coli, and increased both its intrinsic stability and catalytic turnover, without affecting its ligand-binding profile. The partial ordering of the activation loop in the EphB3 structure alludes to a potential cis-phosphorylation mechanism for the EphB kinases. With the kinase domain structures of all four catalytically competent human EphB receptors now determined, a picture begins to emerge of possible opportunities to produce EphB isozyme-selective kinase inhibitors for mechanistic studies and therapeutic applications.


Subject(s)
Receptor, EphB1/chemistry , Receptor, EphB2/chemistry , Receptor, EphB4/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Mutagenesis , Protein Conformation , Protein Stability , Protein Structure, Tertiary , Receptor, EphB3/chemistry , Receptor, EphB3/genetics
20.
Biosci Rep ; 33(3)2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23627399

ABSTRACT

The Eph (erythropoietin-producing hepatocellular carcinoma) B receptors are important in a variety of cellular processes through their roles in cell-to-cell contact and signalling; their up-regulation and down-regulation has been shown to have implications in a variety of cancers. A greater understanding of the similarities and differences within this small, highly conserved family of tyrosine kinases will be essential to the identification of effective therapeutic opportunities for disease intervention. In this study, we have developed a route to production of multi-milligram quantities of highly purified, homogeneous, recombinant protein for the kinase domain of these human receptors in Escherichia coli. Analyses of these isolated catalytic fragments have revealed stark contrasts in their amenability to recombinant expression and their physical properties: e.g., a >16°C variance in thermal stability, a 3-fold difference in catalytic activity and disparities in their inhibitor binding profiles. We find EphB3 to be an outlier in terms of both its intrinsic stability, and more importantly its ligand-binding properties. Our findings have led us to speculate about both their biological significance and potential routes for generating EphB isozyme-selective small-molecule inhibitors. Our comprehensive methodologies provide a template for similar in-depth studies of other kinase superfamily members.


Subject(s)
Receptors, Eph Family/chemistry , Receptors, Eph Family/metabolism , Amino Acid Sequence , Cloning, Molecular , Enzyme Stability , Escherichia coli/genetics , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Receptors, Eph Family/antagonists & inhibitors , Receptors, Eph Family/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL